Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Biochem Biophys Res Commun ; 440(2): 271-6, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-24055706

ABSTRACT

Vesicular stomatitis virus (VSV) is a prototypic negative sense single-stranded RNA virus. The bullet-shape appearance of the virion results from tightly wound helical turns of the nucleoprotein encapsidated RNA template (N-RNA) around a central cavity. Transcription and replication require polymerase complexes, which include a catalytic subunit L and a template-binding subunit P. L and P are inferred to be in the cavity, however lacking direct observation, their exact position has remained unclear. Using super-resolution fluorescence imaging and atomic force microscopy (AFM) on single VSV virions, we show that L and P are packaged asymmetrically towards the blunt end of the virus. The number of L and P proteins varies between individual virions and they occupy 57 ± 12 nm of the 150 nm central cavity of the virus. Our finding positions the polymerases at the opposite end of the genome with respect to the only transcriptional promoter.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Vesicular stomatitis Indiana virus/genetics , Viral Proteins/metabolism , Virus Assembly/physiology , Microscopy, Atomic Force , Microscopy, Fluorescence , RNA, Viral/genetics , Vesicular stomatitis Indiana virus/ultrastructure
2.
Sci Rep ; 10(1): 10643, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32606395

ABSTRACT

Within 2-6 hours after infection by vesicular stomatitis virus (VSV), newly assembled VSV particles are released from the surface of infected cells. In that time, viral ribonucleoprotein (RNP) particles (nucleocapsids) travel from their initial sites of synthesis near the nucleus to the edge of the cell, a distance of 5-10 µm. The hydrodynamic radius of RNP particles (86 nm) precludes simple diffusion through the mesh of cytoskeletal fibers. To reveal the relative importance of different transport mechanisms, movement of GFP-labeled RNP particles in live A549 cells was recorded within 3 to 4 h postinfection at 100 frames/s by fluorescence video microscopy. Analysis of more than 200 RNP particle tracks by Bayesian pattern recognition software found that 3% of particles showed rapid, directional motion at about 1 µm/s, as previously reported. 97% of the RNP particles jiggled within a small, approximately circular area with Gaussian width σ = 0.06 µm. Motion within such "traps" was not directional. Particles stayed in traps for approximately 1 s, then hopped to adjacent traps whose centers were displaced by approximately 0.17 µm. Because hopping occurred much more frequently than directional motion, overall transport of RNP particles was dominated by hopping over the time interval of these experiments.


Subject(s)
Cytoplasm/virology , Motion , Nucleocapsid/ultrastructure , Vesicular stomatitis Indiana virus/physiology , A549 Cells , Diffusion , Humans , Nucleocapsid/physiology , Single Molecule Imaging , Vesicular stomatitis Indiana virus/ultrastructure
3.
J Virol ; 82(11): 5536-47, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367537

ABSTRACT

Immunogold electron microscopy and analysis were used to determine the organization of the major structural proteins of vesicular stomatitis virus (VSV) during virus assembly. We determined that matrix protein (M protein) partitions into plasma membrane microdomains in VSV-infected cells as well as in transfected cells expressing M protein. The sizes of the M-protein-containing microdomains outside the virus budding sites (50 to 100 nm) were smaller than those at sites of virus budding (approximately 560 nm). Glycoprotein (G protein) and M protein microdomains were not colocalized in the plasma membrane outside the virus budding sites, nor was M protein colocalized with microdomains containing the host protein CD4, which efficiently forms pseudotypes with VSV envelopes. These results suggest that separate membrane microdomains containing either viral or host proteins cluster or merge to form virus budding sites. We also determined whether G protein or M protein was colocalized with VSV nucleocapsid protein (N protein) outside the budding sites. Viral nucleocapsids were observed to cluster in regions of the cytoplasm close to the plasma membrane. Membrane-associated N protein was colocalized with G protein in regions of plasma membrane of approximately 600 nm. In contrast to the case for G protein, M protein was not colocalized with these areas of nucleocapsid accumulation. These results suggest a new model of virus assembly in which an interaction of VSV nucleocapsids with G-protein-containing microdomains is a precursor to the formation of viral budding sites.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Nucleocapsid/metabolism , Vesicular stomatitis Indiana virus/metabolism , Viral Envelope Proteins/metabolism , Viral Matrix Proteins/metabolism , Animals , Cell Line , Cricetinae , Cytoplasm/metabolism , Membrane Microdomains/ultrastructure , Microscopy, Immunoelectron , Vesicular stomatitis Indiana virus/ultrastructure , Viral Matrix Proteins/ultrastructure
4.
J Cell Biol ; 98(4): 1304-19, 1984 Apr.
Article in English | MEDLINE | ID: mdl-6325468

ABSTRACT

Madin-Darby canine kidney (MDCK) cells can sustain double infection with pairs of viruses of opposite budding polarity (simian virus 5 [SV5] and vesicular stomatitis virus [VSV] or influenza and VSV), and we observed that in such cells the envelope glycoproteins of the two viruses are synthesized simultaneously and assembled into virions at their characteristic sites. Influenza and SV5 budded exclusively from the apical plasma membrane of the cells, while VSV emerged only from the basolateral surfaces. Immunoelectron microscopic examination of doubly infected MDCK cells showed that the influenza hemagglutinin (HA) and the VSV G glycoproteins traverse the same Golgi apparatus and even the same Golgi cisternae. This indicates that the pathways of the two proteins towards the plasma membrane do not diverge before passage through the Golgi apparatus and therefore that critical sorting steps must take place during or after passage of the glycoproteins through this organelle. After its passage through the Golgi, the HA accumulated primarily at the apical membrane, where influenza virion assembly occurred. A small fraction of HA did, however, appear on the lateral surface and was incorporated into the envelope of budding VSV virions. Although predominantly found on the basolateral surface, significant amounts of G protein were observed on the apical plasma membrane well before disruption of the tight junctions was detectable. Nevertheless, assembly of VSV virions was restricted to the basolateral domain and in doubly infected cells the G protein was only infrequently incorporated into the envelope of budding influenza virions. These observations indicate that the site of VSV budding is not determined exclusively by the presence of G polypeptides. Therefore, it is likely that, at least for VSV, other cellular or viral components are responsible for the selection of the appropriate budding domain.


Subject(s)
Cell Transformation, Viral , Glycoproteins/metabolism , Golgi Apparatus/physiology , Orthomyxoviridae/genetics , Polyomavirus/genetics , Vesicular stomatitis Indiana virus/genetics , Viral Proteins/metabolism , Animals , Biological Transport , Cell Line , Dogs , Kidney/physiology , Microscopy, Electron , Polyomavirus/ultrastructure , Vesicular stomatitis Indiana virus/ultrastructure
5.
J Cell Biol ; 84(2): 421-9, 1980 Feb.
Article in English | MEDLINE | ID: mdl-6247354

ABSTRACT

The single glycoprotein (G) of vesiclar stomatitis virus (VSV) was isolated in nearly quantitative yield by extraction of the purified virions with 0.05 M octyl-beta-D- glucoside (OG) in 0.01 M sodium phosphate, pH 8.0. The extract contained essentially all of the viral phospholipids and glycolipids, and was free of other essentially all of the viral phospholipids and glycolipids, and was free of other viral proteins. Dialysis to remove OG resulted in the formation of G protein-viral lipid vesicles having a lipid-G protein ratio similar to that of the intact virions. The vesicles were 250-1,000 A in diameter, with a "fuzzy" external layer also similar to that of intact virions. The vesicles were predominantly unilamellar and sealed, with both phosphatidyl ethanolamine and gangliosides symmetrically distributed in the bilayer. G protein was asymmetrically oriented, with about 80 percent accessible to exogenous protease. Addition of soybean phospholipid to the viral extract before dialysis resulted in vesicles that incorporated viral proteins and lipids quantitatively, but that were markedly decreased in buoyant density. The G neutralized protein-lipid vesicles were effective in eliciting specific anti-G antibodies that neutralized viral infectivity. Competitive radioimmunoassay showed that both reconstituted vesicles and a soluble form of G protein (Gs) were indistinguishable from purified VSV in their antibody binding properties. Addition of G protein-lipid vesicles of BHK-21 cells before, or simultaneously with, infection by VSV inhibited viral infectivity, as measured by two independent techniques (viral RNA production in the presence of actinomycin D and a neutral red assay of cell viability). The total inhibitory activity of G protein in the vesicular form was, however, less than 5 percent of that found for intact virus particles that have been inactivated by ultraviolet light irradiation. Gs was inactive as an inhibitor as determined by the RNA production assay.


Subject(s)
Glycoproteins , Lipids , Vesicular stomatitis Indiana virus/ultrastructure , Viral Proteins , Animals , Cell Line , Cricetinae , Cytopathogenic Effect, Viral , Glycoproteins/analysis , Kidney , Lipids/analysis , Protein Conformation , RNA, Viral/biosynthesis , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/metabolism , Viral Proteins/analysis
6.
J Cell Biol ; 97(6): 1777-87, 1983 Dec.
Article in English | MEDLINE | ID: mdl-6315743

ABSTRACT

An immunoelectron microscopic study was undertaken to survey the intracellular pathway taken by the integral membrane protein (G-protein) of vesicular stomatitis virus from its site of synthesis in the rough endoplasmic reticulum to the plasma membrane of virus-infected Chinese hamster ovary cells. Intracellular transport of the G-protein was synchronized by using a temperature-sensitive mutant of the virus (0-45). At the nonpermissive temperature (39.8 degrees C), the G-protein is synthesized in the cell infected with 0-45, but does not leave the rough endoplasmic reticulum. Upon shifting the temperature to 32 degrees C, the G-protein moves by stages to the plasma membrane. Ultrathin frozen sections of 0-45-infected cells were prepared and indirectly immunolabeled for the G-protein at different times after the temperature shift. By 3 min, the G-protein was seen at high density in saccules at one face of the Golgi apparatus. No large accumulation of G-protein-containing vesicles were observed near this entry face, but a few 50-70-mm electron-dense vesicular structures labeled for G-protein were observed that might be transfer vesicles between the rough endoplasmic reticulum and the Golgi complex. At blebbed sites on the nuclear envelope at these early times there was a suggestion that the G-protein was concentrated, these sites perhaps serving as some of the transitional elements for subsequent transfer of the G-protein from the rough endoplasmic reticulum to the Golgi complex. By 3 min after its initial asymmetric entry into the Golgi complex, the G-protein was uniformly distributed throughout all the saccules of the complex. At later times, after the G-protein left the Golgi complex and was on its way to the plasma membrane, a new class of G-protein-containing vesicles of approximately 200-nm diameter was observed that are probably involved in this stage of the transport process. These data are discussed, and the further prospects of this experimental approach are assessed.


Subject(s)
Cell Transformation, Viral , Membrane Glycoproteins , Vesicular stomatitis Indiana virus/ultrastructure , Viral Envelope Proteins , Viral Proteins/metabolism , Animals , Biological Transport , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cricetinae , Cricetulus , Female , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Microscopy, Electron , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Ovary , Vesicular stomatitis Indiana virus/metabolism
7.
J Cell Biol ; 107(2): 471-9, 1988 Aug.
Article in English | MEDLINE | ID: mdl-2458357

ABSTRACT

Caco-2 cells, derived from human colon, have the morphological, functional, and biochemical properties of small intestinal epithelial cells. After infection with enveloped viruses, influenza virions assembled at the apical plasma membrane while vesicular stomatitis virus (VSV) particles appeared exclusively at the basolateral membrane, similar to the pattern observed in virus-infected Madin-Darby canine kidney (MDCK). When grown in Millicell filter chamber devices and labeled with [35S]methionine, Caco-2 monolayers released all of their radiolabeled secretory products preferentially into the basal chamber. Among the proteins identified were apolipoproteins AI and E, transferrin, and alpha-fetoprotein. No proteins were observed to be secreted preferentially from the apical cell surface. The lysosomal enzyme beta-hexosaminidase was also secreted primarily from the basolateral surface of the cells in the presence or absence of lysosomotropic drugs or tunicamycin, which inhibit the targetting of lysosomal enzymes to lysosomes. Neither of these drug treatments significantly affected the polarized secretion of other nonlysosomal proteins. In addition, growth hormone (GH), which is released in a nonpolar fashion from MDCK cells, was secreted exclusively from the basolateral membrane after transfection of Caco-2 cells with GH cDNA in a pSV2-based expression vector. Similar results were obtained in transient expression experiments and after selection of permanently transformed Caco-2 cells expressing GH. Since both beta-hexosaminidase and GH would be expected to lack sorting signals for polarized exocytosis in epithelial cells, these results indicate that in intestinal cells, proteins transported via the basolateral secretory pathway need not have specific sorting signals.


Subject(s)
Intestinal Mucosa/metabolism , Protein Biosynthesis , Apolipoproteins/biosynthesis , Autoradiography , Carcinoma , Colonic Neoplasms , Electrophoresis, Polyacrylamide Gel , Epithelium/enzymology , Epithelium/metabolism , Epithelium/microbiology , Growth Substances/biosynthesis , Humans , Intestines/enzymology , Intestines/microbiology , Microscopy, Electron , Orthomyxoviridae/physiology , Orthomyxoviridae/ultrastructure , Precipitin Tests , Transferrin/biosynthesis , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/physiology , Vesicular stomatitis Indiana virus/ultrastructure , Virion/physiology , Virion/ultrastructure , alpha-Fetoproteins/biosynthesis , beta-N-Acetylhexosaminidases/biosynthesis
8.
J Virol Methods ; 153(1): 74-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18598719

ABSTRACT

It is well documented in the scientific literature that ozone-oxygen mixtures inactivate microorganisms including bacteria, fungi and viruses (Hoff, J.C., 1986. Inactivation of microbial agents by chemical disinfectants. EPA 600 S2-86 067. Office of Water, U.S. Environmental Protection Agency, Washington, DC; Khadre, M.A., Yousef, A.E., Kim, J.-G., 2001. Microbiological aspects of ozone applications in food: a review. J. Food Sci. 66, 1242-1252). In the current study, delivery and absorption of precisely known concentrations of ozone (in liquid media) were used to inactivate virus infectivity. An ozone-oxygen delivery system capable of monitoring and recording ozone concentrations in real time was used to inactivate a series of enveloped and non-enveloped viruses including herpes simplex virus type-1 (HHV-1, strain McIntyre), vesicular stomatitis Indiana virus (VSIV), vaccinia virus (VACV, strain Elstree), adenovirus type-2 (HAdV-2), and the PR8 strain of influenza A virus (FLUAVA/PR/8/34/H1N1; FLUAV). The results of the study showed that ozone exposure reduced viral infectivity by lipid peroxidation and subsequent lipid envelope and protein shell damage. These data suggest that a wide range of virus types can be inactivated in an environment of known ozone exposure.


Subject(s)
Disinfectants/pharmacology , Ozone/pharmacology , Reactive Oxygen Species/pharmacology , Virion/drug effects , Virus Inactivation , Adenoviridae/drug effects , Adenoviridae/ultrastructure , Influenza A virus/drug effects , Influenza A virus/ultrastructure , Microscopy, Electron, Transmission , Simplexvirus/drug effects , Simplexvirus/ultrastructure , Vaccinia virus/drug effects , Vaccinia virus/ultrastructure , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/ultrastructure , Viral Plaque Assay , Virion/ultrastructure
9.
J Virol Methods ; 228: 10-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26562056

ABSTRACT

Only very few comparative studies have been performed that evaluate general trends of virus growth under 3D in comparison with 2D cell culture conditions. The aim of this study was to investigate differences when four animal viruses are cultured in 2D and 3D. Suid herpesvirus 1 (SuHV-1), Vesicular stomatitis virus (VSIV), Bovine adenovirus (BAdV) and Bovine parainfluenza 3 virus (BPIV-3) were cultivated in 3D rotating wall vessels (RWVs) and conventional 2D cultures. The production of virus particles, the portion of infectious particles, and the infectious growth curves were compared. For all viruses, the production of virus particles (related to cell density), including the non-infectious ones, was lower in 3D than in 2D culture. The production of only infectious particles was significantly lower in BAdV and BPIV-3 in 3D cultures in relation to cell density. The two cultivation approaches resulted in significantly different virus particle-to-TCID50 ratios in three of the four viruses: lower in SuHV-1 and BPIV-3 and higher in BAdV in 3D culture. The infectious virus growth rates were not significantly different in all viruses. Although 3D RWV culture resulted in lower production of virus particles compared to 2D systems, the portion of infectious particles was higher for some viruses.


Subject(s)
Atadenovirus/growth & development , Cell Culture Techniques , Herpesvirus 1, Suid/growth & development , Parainfluenza Virus 3, Bovine/growth & development , Vesicular stomatitis Indiana virus/growth & development , Virus Cultivation/methods , Animals , Atadenovirus/physiology , Atadenovirus/ultrastructure , Cattle , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Chlorocebus aethiops , Dogs , Herpesvirus 1, Suid/physiology , Herpesvirus 1, Suid/ultrastructure , Madin Darby Canine Kidney Cells , Parainfluenza Virus 3, Bovine/physiology , Parainfluenza Virus 3, Bovine/ultrastructure , Swine , Vero Cells , Vesicular stomatitis Indiana virus/physiology , Vesicular stomatitis Indiana virus/ultrastructure , Virus Replication
10.
Biochim Biophys Acta ; 464(1): 234-44, 1977 Jan 04.
Article in English | MEDLINE | ID: mdl-188470

ABSTRACT

The proton decoupled 40.48 M Hz 31P NMR spectrum of intact and unperturbed membrane-enclosed vesicular stomatitis virus (sterotype Indiana) exhibited two distinct maxima. These can be resolved into a narrow, symmetric line and a broad asymmetric line. The 31P NMR spectrum of a multilamellar (unsonicated) preparation of the extracted viral lipids exhibited a line shape similar to that of the intact virus. A sonicated vesicle preparation of the extracted viral lipids exhibited a narrow symmetric line. The narrow component in the intact virus spectrum may be attributed to small membrane fragments. Phospholipase C digestion of the intact virus resulted in substantial reduction in intensity of both components which suggests that much of the contribution to both peaks is due to phosphate in the phospholipid polar head groups. The phospholipid phosphates in both sonicated and unsonicated preparations of the extracted viral lipids exhibited substantially longer relaxation times than did those in the intact virus. The short relaxation time emanating from the intact virus preparation is caused by immobilization of the phospholipid head groups which could be due to lipid-protein interactions. Trypsin treatment of vesicular stomatitis virions, which results in complete removal of the exterior hydrophilic segment of the membrane glycoprotein, increased the 31P relaxation time to a value similar to that observed in the protein-free total lipid extracts; this finding provides supporting evidence for the role of virus glycoprotein in shortened relaxation times. A reversible temperature-dependent change in apparent line width and absence of an effect of cholesterol on the 31P phospholipid spectrum were also demonstrated.


Subject(s)
Vesicular stomatitis Indiana virus/ultrastructure , Cell Line , Cholesterol , Magnetic Resonance Spectroscopy , Membrane Lipids/analysis , Membranes/ultrastructure , Microscopy, Electron , Phospholipases , Phosphorus Isotopes
11.
Biochim Biophys Acta ; 433(1): 63-74, 1976 Apr 16.
Article in English | MEDLINE | ID: mdl-177082

ABSTRACT

Observations of the light-scattering properties of several enveloped viruses indicate that virions (vesicular stomatitis, SV5 and influenza), in common with other membrane systems, are osmotically active, responding to NaCl gradients by swelling in hypo-osmolar solutions and shrinking in hyperosmolar solutions. The permeability barrier responsible for this osmotic response in vesicular stomatitis virions was modified both by protease treatment to remove the viral glycoprotein and by treatment with the polyene antibiotic filipin, an agent known to interact with cholesterol in liposomes and membranes. Filipin altered the kinetic and equilibrium permeability behavior of virions but the extent of leakage of osmotic shocking agent was less than that in lecithin/cholesterol and lecithin/ergosterol liposomes and in ergosterol-containing ciliary membranes. Negative-staining electron microscopy revealed that filipin treatment caused structural changes in the viral membrane. Intact virions exhibited appreciably larger responses to osmotic change than did protease-treated virus particles. Thus, the osmotic barrier in intact vesicular stomatitis virions may not be exclusively lipid in nature.


Subject(s)
Membranes/metabolism , Vesicular stomatitis Indiana virus/metabolism , Kinetics , Light , Mathematics , Membranes/ultrastructure , Microscopy, Electron , Osmosis , Permeability , Scattering, Radiation , Vesicular stomatitis Indiana virus/ultrastructure , Viral Proteins/metabolism , Water/metabolism
12.
Biomaterials ; 26(34): 7173-82, 2005 Dec.
Article in English | MEDLINE | ID: mdl-15955554

ABSTRACT

We report a study that investigates the biocompatibility of materials that form lyotropic liquid crystals (LCs) with viruses and mammalian cells that support the replication of viruses. This study is focused on aqueous solutions of tetradecyldimethyl-amineoxide (C(14)AO) and decanol (D), or disodium cromoglycate (DSCG; C(23)H(14)O(11)Na(2)), which can form optically birefringent, liquid crystalline phases. The influence of these materials on the ability of vesicular stomatitis virus (VSV) to infect human epitheloid cervical carcinoma (HeLa) cells was examined by two approaches. First, VSV was dispersed in aqueous C(14)AO+ D or DSCG, and then HeLa cells were inoculated by contacting the cells with the aqueous C(14)AO + D or DSCG containing VSV. The infectivity of VSV to the HeLa cells was subsequently determined. Second, VSV was incubated in LC phases of either C(14)AO + D or DSCG for 4 h, and the concentration (titer) of infectious virus in the LC was determined by dilution into cell culture medium and subsequent inoculation of HeLa cells. Using these approaches, we found that the LC containing C(14)AO + D caused inactivation of virus as well as cell death. In contrast, we determined that VSV retained its infectivity in the presence of aqueous DSCG, and that greater than 74-82% of the HeLa cells survived contact with aqueous DSCG (depending on concentration of DSCG). Because VSV maintained its function (and we infer structure) in LCs formed from DSCG, we further explored the influence of the virus on the ordering of the LC. Whereas the LC formed from DSCG was uniformly aligned on surfaces prepared from self-assembled monolayers (SAMs) of HS(CH(2))(11)(OCH(2)CH(2))(4)OH on obliquely deposited films of gold in the absence of VSV, the introduction of 10(7)-10(8) infectious virus particles per milliliter caused the LC to assume a non-uniform orientation and a colorful appearance that was readily distinguished from the uniformly aligned LCs. Control experiments using cell lysates with equivalent protein concentrations but no virus did not perturb the uniform alignment of the LC.


Subject(s)
Biocompatible Materials/chemistry , Crystallization/methods , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/ultrastructure , Virus Cultivation/methods , Virus Replication/physiology , HeLa Cells , Humans , Materials Testing , Solutions
13.
Hum Gene Ther ; 8(17): 2011-8, 1997 Nov 20.
Article in English | MEDLINE | ID: mdl-9414250

ABSTRACT

Baculovirus vectors recently have been shown to be capable of efficient transduction of human hepatoma cells and primary hepatocytes in culture. This paper describes the generation of a novel recombinant baculovirus (VGZ3) in which the vesicular stomatitis virus glycoprotein G (VSV G) is present in the viral envelope. The gene encoding VSV G was inserted into the baculovirus genome under the control of the polyhedrin promoter such that it was expressed at very high levels in infected insect cells but not in mammalian cells. Expression of the lacZ reporter gene was driven by a promoter that is functional in mammalian cells (the Rous sarcoma virus long terminal repeat). We show by Western analysis that VSV G protein was present in purified baculovirus preparations. A VSV G monoclonal antibody blocked transduction of mammalian cells by VGZ3. This virus was morphologically distinct from baculovirus lacking VSV G, with virions adopting an oval rather than rod-shaped morphology. VGZ3 transduced human hepatoma cells in vitro at an efficiency roughly 10-fold greater than baculovirus lacking VSV G (the virus Z4). VGZ3 was also capable of transducing cell lines that could not be transduced efficiently by Z4. We provide evidence that VSV G protein may enhance transduction by increasing the efficiency of escape of baculovirus from intracellular vesicles rather than by increasing cell binding or uptake of the virus. The possible use of this and related baculoviruses in gene therapy is discussed.


Subject(s)
Baculoviridae/genetics , Genetic Vectors/genetics , Membrane Glycoproteins , Transformation, Genetic , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Animals , Baculoviridae/ultrastructure , Cell Line , Cell Nucleus/virology , Chlorocebus aethiops , Gene Transfer Techniques , Humans , Lac Operon , Mice , Spodoptera/cytology , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/ultrastructure
14.
Biochimie ; 72(4): 235-43, 1990 Apr.
Article in English | MEDLINE | ID: mdl-1696507

ABSTRACT

The role of N2-methyl-9-hydroxy-ellipticine (NMHE) and N2,N6-dimethyl-9-hydroxy-ellipticine (DMHE) in modulating the antiviral activity of poly (A-U) was examined using a human foreskin fibroblast-vesicular stomatitis virus (HSF-VSV) bioassay in which the concentration of poly (A-U) was fixed at 0.05 mM or 0.2 mM while the NMHE or DMHE concentration was varied to produce variable NMHE (or DMHE)/ribonucleotide ratios ranging from 1:16 to 2:1. Poly (A-U), NMHE and DMHE tested individually were not efficacious antiviral agents. When the poly (A-U) was combined with the NMHE or DMHE, the antiviral activity of the poly (A-U) was potentiated 16- to 20-fold a NMHE (or DMHE)/ribonucleotide ratios in the region of 1/4. Poly (A-U), NMHE and DMHE induce beta-IFN. The interferon-inducing activity of the NMHE (or DMHE)/poly (A-U) combination was equal to the sum of the interferon-inducing activity of the poly (A-U) alone and the NMHE (or DMHE) alone. The direct viral inactivation study demonstrated that NMHE, DMHE, poly (A-U) and the NMHE (or DMHE)/poly (A-U) combinations did not inactivate VSV at concentrations near the 50% viral inhibitory dose. Photomicrographs of HSF cells incubated with NMHE alone or with a NMHE/poly (A-U) combination suggest that poly (A-U) affects the subcellular distribution of the NMHE by steering the NMHE to the nucleolus. These observations suggest that modulation of a nuclear process may be responsible for the enhanced antiviral activity.


Subject(s)
Alkaloids/pharmacology , Antiviral Agents , Ellipticines/pharmacology , Poly A-U/pharmacology , Vesicular stomatitis Indiana virus/drug effects , Cells, Cultured , Fibroblasts/microbiology , Fibroblasts/ultrastructure , Humans , Interferons/pharmacology , Vesicular stomatitis Indiana virus/growth & development , Vesicular stomatitis Indiana virus/ultrastructure , Virus Activation/drug effects
15.
Am J Trop Med Hyg ; 25(1): 177-85, 1976 Jan.
Article in English | MEDLINE | ID: mdl-176893

ABSTRACT

Analysis of infectious virus particles after intrathoracic injection revealed that Aedes aegypti mosquito tissues are capable of supporting the growth of vesicular stomatitis virus (VSV), serotype Indiana. Although all tissues assayed (salivary gland, midgut, diverticulum, malphigian tubules, and ovary) were capable of supporting VSV growth, the salivary gland was the only organ capable of maintaining an appreciable amount of virus for periods longer than 9 days postinfection. Electron microscopic studies of infected tissues showed virus particles consistently within the cell cytoplasm of all organs with no evidence of nuclear involvement. Direct evidence of crystalline formation of VSV in the apical cavities of salivary gland tissue was demonstrated.


Subject(s)
Aedes , Insect Vectors , Stomatitis, Aphthous/transmission , Vesicular stomatitis Indiana virus , Aedes/ultrastructure , Animals , Vesicular stomatitis Indiana virus/ultrastructure , Viral Plaque Assay
16.
Am J Trop Med Hyg ; 26(2): 299-306, 1977 Mar.
Article in English | MEDLINE | ID: mdl-192094

ABSTRACT

The characteristics and ecology of Isfahan virus, a new vesicular stomatitis virus (VSV) serotype, are described. Two strains of the agent were isolated from pools of Phlebotomus papatasi collected in Iran in 1975. Its animal pathogenicity, growth rate, cytopathic effect, and plaque morphology are similar to those of the other VSV serotypes. Electron microscopic examination of the virus demonstrated a bullet shape, the presence of truncated particles and maturation at plasma membranes. Antigenic relationships between Isfahan virus and three other VSV serotypes (Cocal, Piry, and Chandipura) were demonstrated by complement fixation or neutralization tests. A high prevalence of Isfahan neutralizing antibodies was found in human sera from several regions of Iran, suggesting that the virus may be of some public health importance. All of the residents over 5 years of age in the village where the virus was isolated had been infected. Neutralizing antibodies to Isfahan virus were also found in sera of Iranian gerbils but not in domestic animals. Results of this study suggest that the ecology of Isfahan virus is distinct from the other VSV serotypes and involves chiefly humans, gerbils, and sandflies, a pattern also observed with cutaneous leishmaniasis and sandfly fever in Iran.


Subject(s)
Gerbillinae/microbiology , Psychodidae/microbiology , Vesicular stomatitis Indiana virus/physiology , Animals , Antibodies, Viral/analysis , Antigens, Viral , Cattle , Complement Fixation Tests , Cytopathogenic Effect, Viral , Ecology , Female , Humans , Inclusion Bodies, Viral , Mice , Mites/microbiology , Neutralization Tests , Rodent Diseases/immunology , Rodent Diseases/microbiology , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/ultrastructure , Virus Diseases/epidemiology , Virus Diseases/immunology , Virus Diseases/veterinary
17.
J Virol Methods ; 23(1): 1-12, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2536378

ABSTRACT

Spreading of cells on a solution surface could visualize vesicular stomatitis virus nucleocapsids and virions in infected cells easily and clearly without the need for any purification. Characteristic structures observed by the spreading of the infected cells are described and discussed.


Subject(s)
Capsid/ultrastructure , Vesicular stomatitis Indiana virus/ultrastructure , Viral Core Proteins/ultrastructure , Virology/methods , Animals , Cells, Cultured , Microscopy, Electron
18.
J Antibiot (Tokyo) ; 52(7): 613-9, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10513840

ABSTRACT

Inactivation of enveloped viruses (VSV, SFV, and SHV-1) by surfactin lipopeptides was dependent on the hydrophobicity, i.e. the number of carbon atoms of the fatty acid, and on the charge of the peptide moiety as well as on the virus species. Surfactins with fatty acid chains of 13 carbon atoms showed very low antiviral activity in comparison to C14 and C15 isoforms. C15 surfactin monomethyl ester also inactivated SFV which was resistant to the mixture of surfactin isoforms as produced by Bacillus subtilis. In contrast, the dimethyl ester showed no virus-inactivation capacity. Disintegration of viral structures as determined by electron microscopy after inactivation of VSV and SFV was comparable to the titer reduction. The effect of the surfactin isoforms and methyl esters on erythrocyte hemolysis correlated with the virus-inactivation capacity. Surfactins with a fatty acid chain moiety of 15 carbon atoms and one negative charge showed the highest antiviral activity.


Subject(s)
Antiviral Agents/pharmacology , Bacterial Proteins/pharmacology , Hemolysis/drug effects , Peptides, Cyclic , Animals , Bacterial Proteins/chemistry , Cell Line , Cricetinae , Erythrocytes , Esters/chemistry , Esters/pharmacology , Herpesvirus 1, Suid/drug effects , Herpesvirus 1, Suid/ultrastructure , Humans , In Vitro Techniques , Isomerism , Lipopeptides , Lung/metabolism , Lung/virology , Microscopy, Electron , Mink , Semliki forest virus/drug effects , Semliki forest virus/ultrastructure , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/ultrastructure , Virus Replication/drug effects
19.
Am J Vet Res ; 43(4): 565-8, 1982 Apr.
Article in English | MEDLINE | ID: mdl-6176154

ABSTRACT

Human lymphoblastoid interferon (IFN) had an antiviral activity in bovine embryonic kidney cells that resulted in the release of vesicular stomatitis virus (VSV) particles with decreased infectivity. The inhibition was dose dependent and the cells were highly sensitive to human IFN. Examination of the proteins of VSV released from bovine cells after IFN treatment showed a reduction in the glycoprotein. Electron microscopic studies revealed a large number of VSV particles with characteristic spike-like surface projections released from nontreated cells. There was a reduction in the number of mature virions produced in IFN-treated cells and the virions lacked the characteristic surface projections.


Subject(s)
Interferons/pharmacology , Vesicular stomatitis Indiana virus/drug effects , Animals , Cattle , Cell Line , Humans , In Vitro Techniques , Kidney/cytology , Vesicular stomatitis Indiana virus/analysis , Vesicular stomatitis Indiana virus/ultrastructure , Viral Proteins/analysis
20.
Acta Virol ; 27(2): 105-9, 1983 Mar.
Article in English | MEDLINE | ID: mdl-6135328

ABSTRACT

Two xenotropic murine leukaemia virus (XMuLV)-related proteins--a major envelope glycoprotein gp70 and a 90K protein (probably corresponding to the uncleaved envelope precursor)--were expressed on the surface of mouse L cells as demonstrated by lactoperoxidase-catalysed iodination and immunoprecipitation with anti-XMuLV serum. These two proteins out of many labelled cell surface proteins were selectively incorporated into vesicular stomatitis virus (VSV) virions. Significant differences were found in the amounts of labelled XMuLV-related proteins between L cells and two cell lines infected with XMuLV (rabbit SIRC and lamb LKC cells). The two viral antigens represented only a small proportion of radioactivity on L cells. While in XMuLV-infected SIRC and LKC cells, the gp70 was the major labelled surface protein no detectable amounts of XMuLV-related 90K protein or of cell-specific proteins were found in these cells.


Subject(s)
Antigens, Viral , L Cells/metabolism , Leukemia Virus, Murine/immunology , Vesicular stomatitis Indiana virus/growth & development , Viral Proteins/metabolism , Animals , Antigens, Surface , Cell Line , Humans , Mice , Morphogenesis , Protein Precursors/metabolism , Rabbits , Sheep , Vesicular stomatitis Indiana virus/ultrastructure , Viral Envelope Proteins , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL